Healthy Ageing

A cross disciplinary approach to understanding determinants of healthy ageing, from basic mechanisms to senotherapies, providing a holistic approach to health span.

Staff

Publications

2024

Tokatlidis, K. (2024) MIMAS is a new giant multifunctional player in the mitochondrial megacomplex playground. Cell Reports, 43, (doi: 10.1016/j.celrep.2024.113874)

Minnis, H., van Harmelen, A.-L., Gajwani, R., Rizeq, J., Combet, E., Reynolds, R. M., Gillberg, C., Henderson, M., Ho, F. K., Mondelli, V., Pell, J., Smith, J., Shiels, P. G. (2024) The bio-exposome: intracellular processes, stress physiology and the environment. Nature Mental Health, 2, pp. 132-140. (doi: 10.1038/s44220-023-00180-3)

2023

Neytchev, O., Erlandsson, H., Witasp, A., Nordfors, L., Qureshi, A. R., Iseri, K., Morohoshi, H., Selman, C., Ebert, T., Kublickiene, K., Stenvinkel, P., Shiels, P. G. (2023) Epigenetic clocks indicate that kidney transplantation and not dialysis mitigate the effects of renal ageing. Journal of Internal Medicine, 295, pp. 79-90. (doi: 10.1111/joim.13724)

Catterson, J. H., Minkley, L., Aspe, S., Judd-Mole, S., Moura, S., Dyson, M. C., Rajasingam, A., Woodling, N. S., Atilano, M. L., Ahmad, M., Durrant, C. S., Spires-Jones, T. L., Partridge, L. (2023) Protein retention in the endoplasmic reticulum rescues Aβ toxicity in Drosophila. Neurobiology of Aging, 132, pp. 154-174. (doi: 10.1016/j.neurobiolaging.2023.09.008)

Gould, E. et al. (2023) Same data, different analysts: variation in effect sizes due to analytical decisions in ecology and evolutionary biology. EcoEvoRxiv, (doi: 10.32942/X2GG62)

Sannino, D. R., Dobson, A. J. (2023) Acetobacter pomorum in the Drosophila gut microbiota buffers against host metabolic impacts of dietary preservative formula and batch variation in dietary yeast. Applied and Environmental Microbiology, 89, (doi: 10.1128/aem.00165-23)

Craven, H., Erlandsson, H., McGuinness, D., McGuinness, D., Mafra, D., Ijaz, U. Z., Bergman, P., Shiels, P. G., Stenvinkel, P. (2023) A normative microbiome is not restored following kidney transplantation. Clinical Science, 137, pp. 1563-1575. (doi: 10.1042/CS20230779)

Sahota, V. K., Stone, A., Woodling, N. S., Spiers, J. G., Steinert, J. R., Partridge, L., Augustin, H. (2023) Plum modulates Myoglianin and regulates synaptic function in D. melanogaster. Open Biology, 13, (doi: 10.1098/rsob.230171)

Dobson, A., Voigt, S., Kumpitsch, L., Langer, L., Voigt, E., Ibrahim, R., Dowling, D., Reinhardt, K. (2023) Mitonuclear interactions shape both direct and parental effects of diet on fitness, and involve a SNP in mitoribosomal 16s rRNA. PLoS Biology, 21, (doi: 10.1371/journal.pbio.3002218)

Zhou, Z., Lo, C. K.M., Chan, K. L., Chung, R. S.Y., Pell, J. P., Minnis, H., Shiels, P. G., Ip, P., Ho, F. K. (2023) Child maltreatment and telomere length in middle and older age: retrospective cohort study of 141 748 UK Biobank participants. British Journal of Psychiatry, 223, pp. 377-381. (doi: 10.1192/bjp.2023.33)

Shiels, P. G. (2023) ‘Debugging’ heart failure. Journal of Internal Medicine, (doi: 10.1111/joim.13691)

Panagiotou, N., McGuinness, D., Jaminon, A. M.G., Mees, B., Selman, C., Schurgers, L., Shiels, P. G. (2023) Microvesicle-mediated tissue regeneration mitigates the effects of cellular ageing. Cells, 12, (doi: 10.3390/cells12131707)

Kalogeropoulu, S. K., Rauch-Schmücking, H., Lloyd, E. J., Stenvinkel, P., Shiels, P. G., Johnson, R. J., Fröbert, O., Redtenbacher, I., Burgener, I. A., Painer-Gigler, J. (2023) Formerly bile-farmed bears as a model of accelerated ageing. Scientific Reports, 13, (doi: 10.1038/s41598-023-36447-z)

Baptista, B. G., Fanton, S., Ribeiro, M., Cardozo, L. F.M.F., Regis, B., Alvarenga, L., Ribeiro-Alves, M., Berretta, A. A., Shiels, P. G., Mafra, D. (2023) The effect of Brazilian green propolis extract on inflammation in patients with chronic kidney disease on peritoneal dialysis: a randomised double-blind controlled clinical trial. Phytomedicine, 114, (doi: 10.1016/j.phymed.2023.154731)

Dai, L., Mafra, D., Shiels, P. G., Hackeng, T. M., Stenvinkel, P., Schurgers, L. J. (2023) Vitamin K and hallmarks of ageing: focus on diet and gut microbiome. Nutrients, 15, (doi: 10.3390/nu15122727)

Hill, C., Duffy, S., Kettyle, L. M., McGlynn, L., Sandholm, N., Salem, R. M., Thompson, A., Swan, E. J., Kilner, J., Rossing, P., Shiels, P. G., Lajer, M., Groop, P.-H., Maxwell, A. P., McKnight, A. J. (2023) Differential methylation of telomere-related genes is associated with kidney disease in individuals with type 1 diabetes. Genes, 14, (doi: 10.3390/genes14051029)

Shiels, P., Tran, N., McCavitt, J., Neytchev, O., Stenvinkel, P. (2023) Chronic kidney disease and the exposome of ageing. Springer

Hobson, S., Arefin, S., Witasp, A., Hernandez, L., Kublickiene, K., Shiels, P.G., Stenvinkel, P. (2023) Accelerated vascular aging in chronic kidney disease: the potential for novel therapies. Circulation Research, 132, pp. 950-969. (doi: 10.1161/CIRCRESAHA.122.321751)

Vitale, M., Sanz, A., Scialò, F. (2023) Mitochondrial redox signaling: a key player in aging and disease. Aging, (doi: 10.18632/aging.204659)

Castejon-Vega, B., Cordero, M. D., Sanz, A. (2023) How the disruption of mitochondrial redox signalling contributes to ageing. Antioxidants, 12, (doi: 10.3390/antiox12040831)

Fullerton, J., Bettencourt, C., Daniels, M., Mclean, F., Simpson, S., Smith, A., Woodling, N., Kerr, F. (2023) Creating connections: developing an online space for cross-regional mentorship and network building in the dementia research field. Health Open Research, 4, (doi: 10.12688/amrcopenres.13091.2)

Moreira, L. d. S. G., Brum, I. d. S. d. C., de Vargas Reis, D. C.M., Trugilho, L., Chermut, T. R., Esgalhado, M., Cardozo, L. F.M.F., Stenvinkel, P., Shiels, P. G., Mafra, D. (2023) Cinnamon: an aromatic condiment applicable to chronic kidney disease. Kidney Research and Clinical Practice, 42, pp. 4-26. (doi: 10.23876/j.krcp.22.111)

Chan, K. L., Lo, C. K.M., Chen, X.-Y., Ip, P., Leung, W. C., Shiels, P. G., Pell, J. P., Minnis, H., Ho, F. K. (2023) Association between intimate partner violence and leukocyte telomere length: a retrospective cohort study of 144,049 UK Biobank participants. Epidemiology and Psychiatric Sciences, 32, (doi: 10.1017/S2045796023000112)

2022

Martínez Corrales, G., Li, M., Svermova, T., Goncalves, A., Voicu, D., Dobson, A. J., Southall, T. D., Alic, N. (2022) Transcriptional memory of dFOXO activation in youth curtails later-life mortality through chromatin remodelling and Xbp1. Nature Aging, 2, pp. 1176-1190. (doi: 10.1038/s43587-022-00312-x)

Kataura, T. et al. (2022) Autophagy promotes cell survival by maintaining NAD levels. Developmental Cell, 57, pp. 2584-2598.e11. (doi: 10.1016/j.devcel.2022.10.008)

Mafra, D., Ribeiro, M., Fonseca, L., Regis, B., Cardozo, L. F. M. F., Fragoso Dos Santos, H., Emiliano de Jesus, H., Schultz, J., Shiels, P. G., Stenvinkel, P., Rosado, A. (2022) Archaea from the gut microbiota of humans: Could be linked to chronic diseases? Anaerobe, 77, (doi: 10.1016/j.anaerobe.2022.102629)

Fullerton, J., Bettencourt, C., Daniels, M., Mclean, F., Simpson, S., Smith, A., Woodling, N., Kerr, F. (2022) Creating connections: developing an online space for cross-regional mentorship and network building in the dementia research field. AMRC Open Research, 4, (doi: 10.12688/amrcopenres.13091.1)

Mafra, D., Ugochukwu, S. A., Borges, N. A., Cardozo, L. F. M. F., Stenvinkel, P., Shiels, P. G. (2022) Food for healthier aging: power on your plate. Critical Reviews in Food Science and Nutrition, (doi: 10.1080/10408398.2022.2107611)

Ribeiro, M., Alvarenga, L., Cardozo, L. F. M. F., Kemp, J. A., Lima, L. S., Almeida, J. S. d., Leal, V. d. O., Stenvinkel, P., Shiels, P. G., Mafra, D. (2022) The magical smell and taste: Can coffee be good to patients with cardiometabolic disease? Critical Reviews in Food Science and Nutrition, (doi: 10.1080/10408398.2022.2106938)

Ebert, T., Tran, N., Schurgers, L., Stenvinkel, P., Shiels, P. G. (2022) Ageing – oxidative stress, PTMs and disease. Molecular Aspects of Medicine, 86, (doi: 10.1016/j.mam.2022.101099)

Caccavale, E., Johnson, M. P., Brijbassi, S., Andreazza, A. C., Tokatlidis, K. (2022) Mitochondria and Us: from exploration to global collective. Biochemist, 44, pp. 22-28. (doi: 10.1042/bio_2022_122)

Graham, C., Stefanatos, R., Yek, A. E.H., Spriggs, R. V., Loh, S. H.Y., Huerta Uribe, A., Zhang, T., Martins, L. M., Maddocks, O. D.K., Scialo, F., Sanz, A. (2022) Mitochondrial ROS signalling requires uninterrupted electron flow and is lost during ageing in flies. GeroScience, 44, pp. 1961-1974. (doi: 10.1007/s11357-022-00555-x)

Mafra, D., Cardozo, L., Ribeiro-Alves, M., Bergman, P., Shiels, P. G., Stenvinkel, P. (2022) Short Report: Choline plasma levels are related to Nrf2 transcriptional expression in chronic kidney disease? Clinical Nutrition ESPEN, 50, pp. 318-321. (doi: 10.1016/j.clnesp.2022.06.008)

Ledahawsky, L. M., Terzenidou, M. E., Edwards, R., Kline, R. A., Graham, L. C., Eaton, S. L., van der Hoorn, D., Chaytow, H., Huang, Y.‐T., Groen, E. J.N., Motyl, A. A. L., Lamont, D. J., Tokatlidis, K., Wishart, T. M., Gillingwater, T. H. (2022) The mitochondrial protein Sideroflexin 3 (SFXN3) influences neurodegeneration pathways in vivo. FEBS Journal, 289, pp. 3894-3914. (doi: 10.1111/febs.16377)

Mafra, D., Borges, N. A., Alvarenga, L., Ribeiro, M., Fonseca, L., Leal, V. O., Shiels, P. G., Stenvinkel, P. (2022) Fermented food: should patients with cardiometabolic diseases go back to an early neolithic diet? Critical Reviews in Food Science and Nutrition, (doi: 10.1080/10408398.2022.2077300)

de Souza Gouveia Moreira, L., Fanton, S., Cardozo, L., Borges, N. A., Combet, E., Shiels, P. G., Stenvinkel, P., Mafra, D. (2022) Pink pressure: beetroot (Beta vulgaris rubra) as a possible novel medical therapy for chronic kidney disease. Nutrition Reviews, 80, pp. 1041-1061. (doi: 10.1093/nutrit/nuab074)

Witasp, A., Luttropp, K., Qureshi, A. R., Barany, P., Heimbürger, O., Wennberg, L., Ekström, T. J., Shiels, P. G., Stenvinkel, P., Nordfors, L. (2022) Longitudinal genome-wide DNA methylation changes in response to kidney failure replacement therapy. Scientific Reports, 12, (doi: 10.1038/s41598-021-04321-5)

2021

Tokatlidis, K., Eaglesfield, R. (2021) Targeting and insertion of membrane proteins in mitochondria. Frontiers in Cell and Developmental Biology, 9, (doi: 10.3389/fcell.2021.803205)

Cardozo, L. F.M.F., Alvarenga, L. A., Ribeiro, M., Dai, L., Shiels, P. G., Stenvinkel, P., Lindholm, B., Mafra, D. (2021) Cruciferous vegetables: rationale for exploring potential salutary effects of sulforaphane-rich foods in patients with chronic kidney disease. Nutrition Reviews, 79, pp. 1204-1224. (doi: 10.1093/nutrit/nuaa129)

Castejón-Vega, B., Rubio, A., Pérez-Pulido, A. J., Quiles, J. L., Lane, J. D., Fernández-Domínguez, B., Cachón-González, M. B., Martín-Ruiz, C., Sanz, A., Cox, T. M., Alcocer-Gómez, E., Cordero, M. D. (2021) L-arginine ameliorates defective autophagy in GM2 gangliosidoses by mTOR modulation. Cells, 10, (doi: 10.3390/cells10113122)

Kemp, J. A., Paiva, B. R., dos Santos, H. F., Jesus, H. E., Craven, H., Ijaz, U. Z., Borges, N. A., Shiels, P. G., Mafra, D. (2021) The impact of enriched resistant starch type‐2 cookies on the gut microbiome in hemodialysis patients: a randomized controlled trial. Molecular Nutrition and Food Research, 65, (doi: 10.1002/mnfr.202100374)

Bonfini, A., Dobson, A. J., Duneau, D., Revah, J., Liu, X., Houtz, P., Buchon, N. (2021) Multiscale analysis reveals that diet-dependent midgut plasticity emerges from alterations in both stem cell niche coupling and enterocyte size. eLife, 10, (doi: 10.7554/elife.64125)

Geldon, S., Fernandez-Vizarra, E., Tokatlidis, K. (2021) Redox-mediated regulation of mitochondrial biogenesis, dynamics and respiratory chain assembly in yeast and human cells. Frontiers in Cell and Developmental Biology, 9, (doi: 10.3389/fcell.2021.720656)

Craven, H., McGuinness, D., Buchanan, S., Galbraith, N., Mcguinness, D. H., Jones, B., Combet, E., Mafra, D., Bergman, P., Ellaway, A., Stenvinkel, P., Ijaz, U. Z., Shiels, P. G. (2021) Socioeconomic position links circulatory microbiota differences with biological age. Scientific Reports, 11, (doi: 10.1038/s41598-021-92042-0)

Navas, P., Sanz, A. (2021) Editorial: "Mitochondrial Coenzyme Q Homeostasis: signalling, respiratory chain stability and diseases." Free Radical Biology and Medicine, 169, pp. 12-13. (doi: 10.1016/j.freeradbiomed.2021.04.005)

Ebert, T., Neytchev, O., Witasp, A., Kublickiene, K., Stenvinkel, P., Shiels, P. (2021) Inflammation and oxidative stress in CKD and dialysis patients. Antioxidants and Redox Signaling, 35, pp. 1426-1448. (doi: 10.1089/ars.2020.8184)

Dai, L., Schurgers, L., Shiels, P. G., Stenvinkel, P. (2021) A biomimetic natural sciences approach to understanding the mechanisms of ageing in burden of life style diseases. Clinical Science, 135, pp. 1251-1272. (doi: 10.1042/CS20201452)

Edwards, R., Eaglesfield, R., Tokatlidis, K. (2021) The mitochondrial intermembrane space: the most constricted mitochondrial sub-compartment with the largest variety of protein import pathways. Open Biology, 11, (doi: 10.1098/rsob.210002)

Kooman, J. P., Stenvinkel, P., Shiels, P. G., Feelisch, M., Canaud, B., Kotanko, P. (2021) The oxygen cascade in hemodialysis patients and native high altitude dwellers- lessons from extreme physiology to benefit patients with end-stage renal disease. American Journal of Physiology: Renal Physiology, 320, pp. F249-F261. (doi: 10.1152/ajprenal.00540.2020)

Bolukbasi, E., Woodling, N. S., Ivanov, D. K., Adcott, J., Foley, A., Rajasingam, A., Gittings, L. M., Aleyakpo, B., Niccoli, T., Thornton, J. M., Partridge, L. (2021) Cell type-specific modulation of healthspan by Forkhead family transcription factors in the nervous system. Proceedings of the National Academy of Sciences of the United States of America, 118, (doi: 10.1073/pnas.2011491118)

Scialo, F., Sanz, A. (2021) Coenzyme Q redox signalling and longevity. Free Radical Biology and Medicine, 164, pp. 187-205. (doi: 10.1016/j.freeradbiomed.2021.01.018)

Stenvinkel, P., Painer, J., Shiels, P.G., Bansal, A., Fereidouni, S., Natterson-Horowitz, B., Johnson, R.J., Miranda, J.J. (2021) SARS-COV-2 and biomimetics: what saves the planet will save our health. Journal of Internal Medicine, 289, pp. 244-246. (doi: 10.1111/joim.13128)

Navarro-Pando, J. M., Alcocer-Gómez, E., Castejón-Vega, B., Navarro-Villarán, E., Condés-Hervás, M., Mundi-Roldan, M., Muntané, J., Pérez-Pulido, A. J., Bullon, P., Wang, C., Hoffman, H. M., Sanz, A., Mbalaviele, G., Ryffel, B., Cordero, M. D. (2021) Inhibition of the NLRP3 inflammasome prevents ovarian aging. Science Advances, 7, (doi: 10.1126/sciadv.abc7409)

Fanton, S., Cardozo, L. F.M.F., Combet, E., Shiels, P. G., Stenvinkel, P., Vieira, I. O., Narciso, H. R., Schmitz, J., Mafra, D. (2021) The sweet side of dark chocolate for chronic kidney disease patients. Clinical Nutrition, 40, pp. 15-26. (doi: 10.1016/j.clnu.2020.06.039)

Mafra, D., Borges, N. A., Lindholm, B., Shiels, P. G., Evenepoel, P., Stenvinkel, P. (2021) Food as medicine: targeting the uraemic phenotype in chronic kidney disease. Nature Reviews Nephrology, 17, pp. 153-171. (doi: 10.1038/s41581-020-00345-8)

Klionsky, D. J. et al. (2021) Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition) Autophagy, 17, pp. 1-382. (doi: 10.1080/15548627.2020.1797280)

Anoar, S., Woodling, N. S., Niccoli, T. (2021) Mitochondria dysfunction in frontotemporal dementia/amyotrophic lateral sclerosis: lessons from drosophila models. Frontiers in Neuroscience, 15, (doi: 10.3389/fnins.2021.786076)

Dimogkioka, A.-R., Lees, J., Tokatlidis, K., Lacko, E. (2021) Protein import in mitochondria biogenesis: Guided by targeting signals and sustained by dedicated chaperones. RSC Advances, 11, pp. 32476-32493. (doi: 10.1039/D1RA04497D)

Dickson-Murray, E., Nedara, K., Modjtahedi, N., Tokatlidis, K. (2021) The Mia40/CHCHD4 oxidative folding system: Redox regulation and signaling in the mitochondrial intermembrane space. Antioxidants, 10, (doi: 10.3390/antiox10040592)

2020

Bjedov, I., Cochemé, H. M., Foley, A., Wieser, D., Woodling, N. S., Castillo-Quan, J. I., Norvaisas, P., Lujan, C., Regan, J. C., Toivonen, J. M., Murphy, M. P., Thornton, J., Kinghorn, K. J., Neufeld, T. P., Cabreiro, F., Partridge, L. (2020) Fine-tuning autophagy maximises lifespan and is associated with changes in mitochondrial gene expression in drosophila. PLoS Genetics, 16, (doi: 10.1371/journal.pgen.1009083)

Ebert, T., Painer, J., Bergman, P., Qureshi, A. R., Giroud, S., Stalder, G., Kublickiene, K., Göritz, F., Vetter, S., Bieber, C., Fröbert, O., Arnemo, J. M., Zedrosser, A., Redtenbacher, I., Shiels, P. G., Johnson, R. J., Stenvinkel, P. (2020) Insights in the regulation of trimetylamine N-oxide production using a comparative biomimetic approach suggest a metabolic switch in hibernating bears. Scientific Reports, 10, (doi: 10.1038/s41598-020-76346-1)

Markopoulou, P., Panagiotou, N., Li, A., Bueno-Perez, R., Madden, D., Buchanan, S., Fairen-Jimenez, D., Shiels, P. G., Forgan, R. S. (2020) Identifying differing intracellular cargo release mechanisms by monitoring in vitro drug delivery from MOFs in real time. Cell Reports Physical Science, 1, (doi: 10.1016/j.xcrp.2020.100254)

Alvarenga, L., Cardozo, L. F.M.F., Borges, N., Bengt, L., Stenvinkel, P., Shiels, P. G., Fouque, D., Mafra, D. (2020) Can nutritional interventions modulate the activation of the NLRP3 inflammasome in chronic kidney disease? Food Research International, 136, (doi: 10.1016/j.foodres.2020.109306)

Woodling, N. S., Rajasingam, A., Minkley, L. J., Rizzo, A., Partridge, L. (2020) Independent glial subtypes delay development and extend healthy lifespan upon reduced insulin-PI3K signalling. BMC Biology, 18, (doi: 10.1186/s12915-020-00854-9)

Buchanan, S., Combet, E., Stenvinkel, P., Shiels, P.G. (2020) Klotho, ageing and the failing kidney. Frontiers in Endocrinology, 11, (doi: 10.3389/fendo.2020.00560)

Stenvinkel, P., Shiels, P. G., Painer, J., Miranda, J. J., Natterson-Horowitz, B., Johnson, R. J. (2020) A planetary health perspective for kidney disease. Kidney International, 98, pp. 261-265. (doi: 10.1016/j.kint.2020.03.024)

Yu, Y., Niccoli, T., Ren, Z., Woodling, N. S., Aleyakpo, B., Szabadkai, G., Partridge, L. (2020) PICALM rescues glutamatergic neurotransmission, behavioural function and survival in a Drosophila model of Aβ42 toxicity. Human Molecular Genetics, 29, pp. 2420-2434. (doi: 10.1093/hmg/ddaa125)

Reinhardt, C., Arena, G., Nedara, K., Edwards, R., Brenner, C., Tokatlidis, K., Modjtahedi, N. (2020) AIF meets the CHCHD4/Mia40-dependent mitochondrial import pathway. Biochimica et Biophysica Acta: Molecular Basis of Disease, 1866, (doi: 10.1016/j.bbadis.2020.165746)

Arefin, S., Buchanan, S., Hobson, S., Steinmetz, J., Alsalhi, S., Shiels, P. G., Kublickiene, K., Stenvinkel, P. (2020) Nrf2 in early vascular ageing: calcification, senescence and therapy. Clinica Chimica Acta, 505, pp. 108-118. (doi: 10.1016/j.cca.2020.02.026)

Scialò, F., Sriram, A., Stefanatos, R., Spriggs, R. V., Loh, S. H.Y., Martins, L. M., Sanz, A. (2020) Mitochondrial complex I derived ROS regulate stress adaptation in Drosophila melanogaster. Redox Biology, 32, (doi: 10.1016/j.redox.2020.101450)

Edwards, R., Gerlich, S., Tokatlidis, K. (2020) The biogenesis of mitochondrial intermembrane space proteins. Biological Chemistry, 401, pp. 737-747. (doi: 10.1515/hsz-2020-0114)

Woodling, N. S., Aleyakpo, B., Dyson, M. C., Minkley, L. J., Rajasingam, A., Dobson, A. J., Leung, K. H. C., Pomposova, S., Fuentealba, M., Alic, N., Partridge, L. (2020) The neuronal receptor tyrosine kinase Alk is a target for longevity. Aging Cell, 19, (doi: 10.1111/acel.13137)

Ebert, T., Pawelzik, S.-C., Witasp, A., Arefin, S., Hobson, S., Kublickiene, K., Shiels, P. G., Bäck, M., Stenvinkel, P. (2020) Inflammation and premature ageing in chronic kidney disease. Toxins, 12, (doi: 10.3390/toxins12040227)

O'Toole, P.W., Shiels, P. G. (2020) The role of the microbiota in sedentary life style disorders and ageing: lessons from the animal kingdom. Journal of Internal Medicine, 287, pp. 271-282. (doi: 10.1111/joim.13021)

Gray, S. C., Kinghorn, K. J., Woodling, N. S. (2020) Shifting equilibriums in Alzheimer's disease: The complex roles of microglia in neuroinflammation, neuronal survival and neurogenesis. Neural Regeneration Research, 15, pp. 1208-1219. (doi: 10.4103/1673-5374.272571)

Kooman, J. P., Stenvinkel, P., Shiels, P. G. (2020) Fabry disease: a new model of premature aging? Nephron, 144, pp. 1-4. (doi: 10.1159/000503290)

2019

Gubina, N., Naudi, A., Stefanatos, R., Jove, M., Scialo, F., Fernandez-Ayala, D. J., Rantapero, T., Yurkevych, I., Portero-Otin, M., Nykter, M., Lushchak, O., Navas, P., Pamplona, R., Sanz, A., Anderson, R. (2019) Essential physiological differences characterize short- and long-lived strains of Drosophila melanogaster. Journals of Gerontology Series A: Biological Sciences and Medical Sciences, 74, pp. 1835-1843. (doi: 10.1093/gerona/gly143)

de Kok, M. J., McGuinness, D., Shiels, P. G., de Vries, D. K., Nolthenius, J. B. T., Wijermars, L. G., Rabelink, T. J., Verschuren, L., Stevenson, K. S., Kingsmore, D. B., McBride, M., Ploeg, R. J., Bastiaannet, E., Schaapherder, A. F., Lindeman, J. H. (2019) The neglectable impact of delayed graft function on long-term graft survival in kidneys donated after circulatory death associates with superior organ resilience. Annals of Surgery, 270, pp. 877-883. (doi: 10.1097/SLA.0000000000003515)

Pulli, I., Löf, C., Blom, T., Asghar, M.Y., Lassila, T., Bäck, N., Lin, K.-L., Nyström, J.H., Kemppainen, K., Toivola, D.M., Dufour, E., Sanz, A., Cooper, H.M., Parys, J.B., Törnquist, K. (2019) Sphingosine kinase 1 overexpression induces MFN2 fragmentation and alters mitochondrial matrix Ca2+ handling in HeLa cells. Biochimica et Biophysica Acta: Molecular Cell Research, 1866, pp. 1475-1486. (doi: 10.1016/j.bbamcr.2019.06.006)

Shiels, P. G., Buchanan, S., Selman, C., Stenvinkel, P. (2019) Allostatic load and ageing; linking the microbiome and nutrition with age related health. Biochemical Society Transactions, 47, pp. 1165-1172. (doi: 10.1042/BST20190110)

Stenvinkel, P., Shiels, P. G. (2019) Long-lived animals with negligible senescence: clues for ageing research. Biochemical Society Transactions, 47, pp. 1157-1164. (doi: 10.1042/BST20190105)

Dobson, A. J., Boulton-McDonald, R., Houchou, L., Svermova, T., Ren, Z., Subrini, J., Vazquez-Prada, M., Hoti, M., Rodriguez-Lopez, M., Ibrahim, R., Gregoriou, A., Gkantiragas, A., Bähler, J., Ezcurra, M., Alic, N. (2019) Longevity is determined by ETS transcription factors in multiple tissues and diverse species. PLoS Genetics, 15, (doi: 10.1371/journal.pgen.1008212)

Lang, J., McKie, J., Smith, H., McLaughlin, A., Gillberg, C., Shiels, P. G., Minnis, H. (2019) Adverse childhood experiences, epigenetics and telomere length variation in childhood and beyond: a systematic review of the literature. European Child and Adolescent Psychiatry, (doi: 10.1007/s00787-019-01329-1)

Ellaway, A., Dundas, R., Robertson, T., Shiels, P. G. (2019) More miles on the clock: neighbourhood stressors are associated with telomere length in a longitudinal study. PLoS ONE, 14, (doi: 10.1371/journal.pone.0214380)

Edwards, R., Tokatlidis, K. (2019) The yeast voltage-dependent anion channel Porin: more IMPORTant than just metabolite transport. Molecular Cell, 73, pp. 861-862. (doi: 10.1016/j.molcel.2019.02.028)

Acosta-Herrera, M., Kerick, M., González-Serna, D., Wijmenga, C., Franke, A., Gregersen, P. K., Padyukov, L., Worthington, J., Vyse, T. J., Alarcón-Riquelme, M. E., Mayes, M. D., Martin, J., Chee, M. M., Madhok, R., Shiels, P. (2019) Genome-wide meta-analysis reveals shared new loci in systemic seropositive rheumatic diseases. Annals of the Rheumatic Diseases, 78, pp. 311-319. (doi: 10.1136/annrheumdis-2018-214127)

Mafra, D., Esgalhado, M., Borges, N. A., Cardozo, L. F.M.F., Stockler-Pinto, M. B., Craven, H., Buchanan, S. J., Lindholm, B., Stenvinkel, P., Shiels, P. G. (2019) Methyl donor nutrients in chronic kidney disease: impact on the epigenetic landscape. Journal of Nutrition, 149, pp. 372-380. (doi: 10.1093/jn/nxy289)

Ledeganck, K. J., Gielis, E. M., Abramowicz, D., Stenvinkel, P., Shiels, P. G., Van Craenenbroeck, A. H. (2019) MicroRNAs in AKI and kidney transplantation. Clinical Journal of the American Society of Nephrology, 14, pp. 454-468. (doi: 10.2215/CJN.08020718)

Hobson, S., Arefin, S., Kublickiene, K., Shiels, P. G., Stenvinkel, P. (2019) Senescent cells in early vascular ageing and bone disease of chronic kidney disease—a novel target for treatment. Toxins, 11, (doi: 10.3390/toxins11020082)

Purohit, P. K., Edwards, R., Tokatlidis, K., Saini, N. (2019) MiR-195 regulates mitochondrial function by targeting mitofusin-2 in breast cancer cells. RNA Biology, 16, pp. 918-929. (doi: 10.1080/15476286.2019.1600999)

2018

Stevenson, R., Shapter, O., Aitken, E., Stevenson, K., Shiels, P.G., Kingsmore, D.B. (2018) Has the expansion in extended criteria deceased donors led to a different type of delayed graft function and poorer outcomes? Transplantation Proceedings, 50, pp. 3160-3164. (doi: 10.1016/j.transproceed.2018.07.022)

Thompson, K. et al. (2018) OXA 1L mutations cause mitochondrial encephalopathy and a combined oxidative phosphorylation defect. EMBO Molecular Medicine, 10, (doi: 10.15252/emmm.201809060)

McGuinness, D., Mohammed, S., Monaghan, L., Wilson, P. A., Kingsmore, D. B., Shapter, O., Stevenson, K. S., Coley, S. M., Devey, L., Kirkpatrick, R. B., Shiels, P. G. (2018) A molecular signature for delayed graft function. Aging Cell, 17, (doi: 10.1111/acel.12825)

Ellaway, A., Dundas, R., Olsen, J. R., Shiels, P. G. (2018) Perceived neighbourhood problems over time and associations with adiposity. International Journal of Environmental Research and Public Health, 15, (doi: 10.3390/ijerph15091854)

Panagiotou, N., Neytchev, O., Selman, C., Shiels, P. G. (2018) Extracellular vesicles, ageing, and therapeutic interventions. Cells, 7, (doi: 10.3390/cells7080110)

Woodling, N.S., Partridge, L. (2018) Parkinson's disease: mitochondria parked at the ER hit the snooze button. Neuron, 98, pp. 1059-1061. (doi: 10.1016/j.neuron.2018.06.025)

Tokatlidis, K. (2018) Shaping the import system of mitochondria. eLife, 7, (doi: 10.7554/elife.38209)

Cardenas-Rodriguez, M., Chatzi, A., Tokatlidis, K. (2018) Iron–sulfur clusters: from metals through mitochondria biogenesis to disease. Journal of Biological Inorganic Chemistry, 23, pp. 509-520. (doi: 10.1007/s00775-018-1548-6)

Maguire, D., Talwar, D., Shiels, P. G., McMillan, D. (2018) The role of thiamine dependent enzymes in obesity and obesity related chronic disease states: a systematic review. Clinical Nutrition ESPEN, 25, pp. 8-17. (doi: 10.1016/j.clnesp.2018.02.007)

Dobson, A. J., He, X., Blanc, E., Bolukbasi, E., Feseha, Y., Yang, M., Piper, M. D.W. (2018) Tissue-specific transcriptome profiling of Drosophila reveals roles for GATA transcription factors in longevity by dietary restriction. npj Aging and Mechanisms of Disease, 4, (doi: 10.1038/s41514-018-0024-4)

Egea, J. et al. (2018) Corrigendum to “European contribution to the study of ROS: a summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS)” [Redox Biol. 13 (2017) 94–162] Redox Biology, 14, pp. 694-696. (doi: 10.1016/j.redox.2017.10.001)

Stefanatos, R., Sanz, A. (2018) The role of mitochondrial ROS in the aging brain. FEBS Letters, 592, pp. 743-758. (doi: 10.1002/1873-3468.12902)

Sannino, D. R., Dobson, A. J., Edwards, K., Angert, E. R., Buchon, N. (2018) The Drosophila melanogaster gut microbiota provisions thiamine to its host. mBio, 9, (doi: 10.1128/mBio.00155-18)

Carroll, B., Otten, E. G., Manni, D., Stefanatos, R., Menzies, F. M., Smith, G. R., Jurk, D., Kenneth, N., Wilkinson, S., Passos, J. F., Attems, J., Veal, E. A., Teyssou, E., Seilhean, D., Millecamps, S., Eskelinen, E.-L., Bronowska, A. K., Rubinsztein, D. C., Sanz, A., Korolchuk, V. I. (2018) Oxidation of SQSTM1/p62 mediates the link between redox state and protein homeostasis. Nature Communications, 9, (doi: 10.1038/s41467-017-02746-z)

Simone, R. et al. (2018) G-quadruplex-binding small molecules ameliorate C9orf72 FTD/ALS pathology in vitro and in vivo. EMBO Molecular Medicine, 10, pp. 22-31. (doi: 10.15252/emmm.201707850)

Maguire, D., Neytchev, O., Talwar, D., McMillan, D., Shiels, P. (2018) Telomere homeostasis: interplay with magnesium. International Journal of Molecular Sciences, 19, (doi: 10.3390/ijms19010157)

Stenvinkel, P., Painer, J., Kuro-o, M., Lanaspa, M., Arnold, W., Ruf, T., Shiels, P. G., Johnson, R. J. (2018) Novel treatment strategies for chronic kidney disease: insights from the animal kingdom. Nature Reviews Nephrology, 14, pp. 265-284. (doi: 10.1038/nrneph.2017.169)

Lakhdar, R., McGuinness, D., Drost, E. M., Shiels, P. G., Bastos, R., MacNee, W., Rabinovich, R. A. (2018) Role of accelerated ageing in limb muscle wasting of patients with COPD. International Journal of Chronic Obstructive Pulmonary Disease, 13, pp. 1987-1998. (doi: 10.2147/COPD.S155952)

2017

Filer, D., Thompson, M. A., Takhaveev, V., Dobson, A. J., Kotronaki, I., Green, J. W.M., Heinemann, M., Tullet, J. M.A., Alic, N. (2017) RNA polymerase III limits longevity downstream of TORC1. Nature, 552, pp. 263-267. (doi: 10.1038/nature25007)

Lu, L., Johnman, C., McGlynn, L., Mackay, D. F., Shiels, P. G., Pell, J. P. (2017) Association between exposure to second-hand smoke and telomere length: cross-sectional study of 1303 non-smokers. International Journal of Epidemiology, 46, pp. 1978-1984. (doi: 10.1093/ije/dyx212)

Bossini-Castillo, L. et al. (2017) An MIF promoter polymorphism is associated with susceptibility to pulmonary arterial hypertension in diffuse cutaneous systemic sclerosis. Journal of Rheumatology, 44, pp. 1453-1457. (doi: 10.3899/jrheum.161369)

Kooman, J. P., Dekker, M. J., Usvyat, L. A., Kotanko, P., van der Sande, F. M., Schalkwijk, C. G., Shiels, P. G., Stenvinkel, P. (2017) Inflammation and premature aging in advanced chronic kidney disease. American Journal of Physiology: Renal Physiology, 313, pp. F938-F950. (doi: 10.1152/ajprenal.00256.2017)

Egea, J. et al. (2017) European contribution to the study of ROS: a summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS) Redox Biology, 13, pp. 94-162. (doi: 10.1016/j.redox.2017.05.007)

Cardenas-Rodriguez, M., Tokatlidis, K. (2017) Cytosolic redox components regulate protein homeostasis via additional localisation in the mitochondrial intermembrane space. FEBS Letters, 591, pp. 2661-2670. (doi: 10.1002/1873-3468.12766)

Shiels, P. G., McGuinness, D., Eriksson, M., Kooman, J. P., Stenvinkel, P. (2017) The role of epigenetics in renal ageing. Nature Reviews Nephrology, 13, pp. 471-482. (doi: 10.1038/nrneph.2017.78)

Witasp, A., Van Craenenbroeck, A. H., Shiels, P. G., Ekström, T. J., Stenvinkel, P., Nordfors, L. (2017) Current epigenetic aspects the clinical kidney researcher should embrace. Clinical Science, 131, pp. 1649-1667. (doi: 10.1042/CS20160596)

MacPherson, L., Tokatlidis, K. (2017) Protein trafficking in the mitochondrial intermembrane space: mechanisms and links to human disease. Biochemical Journal, 474, pp. 2533-2545. (doi: 10.1042/BCJ20160627)

Vazirpanah, N., Kienhorst, L.B.E., Van Lochem, E., Wichers, C., Rossato, M., Shiels, P.G., Dalbeth, N., Stamp, L.K., Merriman, T.R., Janssen, M., Radstake, T.R.D.J., Broen, J.C.A. (2017) Patients with gout have short telomeres compared with healthy participants: association of telomere length with flare frequency and cardiovascular disease in gout. Annals of the Rheumatic Diseases, 76, pp. 1309-1315. (doi: 10.1136/annrheumdis-2016-210538)

Scialò, F., Fernández-Ayala, D. J., Sanz, A. (2017) Role of mitochondrial reverse electron transport in ROS signaling: potential roles in health and disease. Frontiers in Physiology, 8, (doi: 10.3389/fphys.2017.00428)

Tesseur, I., Nguyen, A., Chang, B., Li, L., Woodling, N.S., Wyss-Coray, T., Luo, J. (2017) Deficiency in neuronal TGF-β signaling leads to nigrostriatal degeneration and activation of TGF-β signaling protects against MPTP neurotoxicity in mice. Journal of Neuroscience, 37, pp. 4584-4592. (doi: 10.1523/JNEUROSCI.2952-16.2017)

Mizielinska, S., Ridler, C.E., Balendra, R., Thoeng, A., Woodling, N.S., Grässer, F.A., Plagnol, V., Lashley, T., Partridge, L., Isaacs, A.M. (2017) Bidirectional nucleolar dysfunction in C9orf72 frontotemporal lobar degeneration. Acta Neuropathologica Communications, 5, (doi: 10.1186/s40478-017-0432-x)

Shiels, P. G., Stenvinkel, P., Kooman, J. P., McGuinness, D. (2017) Circulating markers of ageing and allostatic load: a slow train coming. Practical Laboratory Medicine, 7, pp. 49-54. (doi: 10.1016/j.plabm.2016.04.002)

Kritsiligkou, P., Chatzi, A., Charalampous, G., Mironov, A., Grant, C. M., Tokatlidis, K. (2017) Unconventional targeting of a thiol peroxidase to the mitochondrial intermembrane space facilitates oxidative protein folding. Cell Reports, 18, pp. 2729-2741. (doi: 10.1016/j.celrep.2017.02.053)

Stenvinkel, P., Luttropp, K., McGuinness, D., Witasp, A., Qureshi, A. R., Wernerson, A., Nordfors, L., Schalling, M., Ripsweden, J., Wennberg, L., Söderberg, M., Bárány, P., Olauson, H., Shiels, P. G. (2017) CDKN2A/p16INK4a expression is associated with vascular progeria in chronic kidney disease. Aging, 9, pp. 494-507. (doi: 10.18632/aging.101173)

Dobson, A. J., Chaston, J. M., Douglas, A. E. (2017) Erratum to: The Drosophila transcriptional network is structured by microbiota. BMC Genomics, 18, (doi: 10.1186/s12864-017-3508-x)

Dobson, A. J., Ezcurra, M., Flanagan, C. E., Summerfield, A. C., Piper, M. D.W., Gems, D., Alic, N. (2017) Nutritional programming of lifespan by FOXO inhibition on sugar-rich diets. Cell Reports, 18, pp. 299-306. (doi: 10.1016/j.celrep.2016.12.029)

Manganas, P., MacPherson, L., Tokatlidis, K. (2017) Oxidative protein biogenesis and redox regulation in the mitochondrial intermembrane space. Cell and Tissue Research, 367, pp. 43-57. (doi: 10.1007/s00441-016-2488-5)

2016

Luttropp, K., Nordfors, L., McGuinness, D., Wennberg, L., Curley, H., Quasim, T., Genberg, H., Sönnerborg, I., Sandberg, J., Schalling, M., Qureshi, A. R., Bárány, P., Shiels, P. G., Stenvinkel, P. (2016) Increased telomere attrition following renal transplantation: impact of anti-metabolite therapy. Transplantation Direct, 2, (doi: 10.1097/TXD.0000000000000629)

Panagiotou, N., Davies, R. W., Selman, C., Shiels, P. G. (2016) Microvesicles as vehicles for tissue regeneration: Changing of the guards. Current Pathobiology Reports, 4, pp. 181-187. (doi: 10.1007/s40139-016-0115-5)

Dobson, A. J., Chaston, J. M., Douglas, A. E. (2016) The Drosophila transcriptional network is structured by microbiota. BMC Genomics, 17, (doi: 10.1186/s12864-016-3307-9)

Kinghorn, K.J., Grönke, S., Castillo-Quan, J.I., Woodling, N.S., Li, L., Sirka, E., Gegg, M., Mills, K., Hardy, J., Bjedov, I., Partridge, L. (2016) A Drosophila model of neuronopathic gaucher disease demonstrates lysosomal-autophagic defects and altered mTOR signalling and is functionally rescued by rapamycin. Journal of Neuroscience, 36, pp. 11654-11670. (doi: 10.1523/JNEUROSCI.4527-15.2016)

Nuebel, E., Manganas, P., Tokatlidis, K. (2016) Orphan proteins of unknown function in the mitochondrial intermembrane space proteome: new pathways and metabolic cross-talk. Biochimica et Biophysica Acta: Molecular Cell Research, 1863, pp. 2613-2623. (doi: 10.1016/j.bbamcr.2016.07.004)

Antoniewski, C., Scialo, F., Sriram, A., Stefanatos, R., Sanz, A. (2016) Practical recommendations for the use of the GeneSwitch Gal4 system to knock-down genes in Drosophila melanogaster. PLoS ONE, 11, (doi: 10.1371/journal.pone.0161817)

Sanz, A. (2016) Mitochondrial reactive oxygen species: Do they extend or shorten animal lifespan? Biochimica et Biophysica Acta: Bioenergetics, 1857, pp. 1116-1126. (doi: 10.1016/j.bbabio.2016.03.018)

Kalef-Ezra, E., Kotzamani, D., Zaganas, I., Katrakili, N., Plaitakis, A., Tokatlidis, K. (2016) Import of a major mitochondrial enzyme depends on synergy between two distinct helices of its presequence. Biochemical Journal, 473, pp. 2813-2829. (doi: 10.1042/BCJ20160535)

Woodling, N.S., Colas, D., Wang, Q., Minhas, P., Panchal, M., Liang, X., Mhatre, S.D., Brown, H., Ko, N., Zagol-Ikapitte, I., Van Der Hart, M., Khroyan, T.V., Chuluun, B., Priyam, P.G., Milne, G.L., Rassoulpour, A., Boutaud, O., Manning-Bog, A.B., Heller, H.C., Andreasson, K.I. (2016) Cyclooxygenase inhibition targets neurons to prevent early behavioural decline in Alzheimer's disease model mice. Brain, 139, pp. 2063-2081. (doi: 10.1093/brain/aww117)

McGuinness, D., Anthony, D. F., Moulisova, V., MacDonald, A. I., Macintyre, A., Thomson, J., Nag, A., Davies, R. W., Shiels, P. G. (2016) Microvesicles but not exosomes from Pathfinder Cells stimulate functional recovery of the pancreas in a mouse streptozotocin-induced diabetes model. Rejuvenation Research, 19, pp. 223-232. (doi: 10.1089/rej.2015.1723)

Johnston, P. R., Dobson, A. J., Rolff, J. (2016) Genomic signatures of experimental adaptation to antimicrobial peptides in Staphylococcus aureus. G3: Genes, Genomes, Genetics, 6, pp. 1535-1539. (doi: 10.1534/g3.115.023622)

Chatzi, A., Manganas, P., Tokatlidis, K. (2016) Oxidative folding in the mitochondrial intermembrane space: a regulated process important for cell physiology and disease. Biochimica et Biophysica Acta: Molecular Cell Research, 1863, pp. 1298-1306. (doi: 10.1016/j.bbamcr.2016.03.023)

Makarova, O., Rodriguez-Rojas, A., Eravci, M., Weise, C., Dobson, A., Johnston, P., Rolff, J. (2016) Antimicrobial defence and persistent infection in insects revisited. Philosophical Transactions of the Royal Society B: Biological Sciences, 371, (doi: 10.1098/rstb.2015.0296)

McClelland, R., Christensen, K., Mohammed, S., McGuinness, D., Cooney, J., Bakhshi, A., Demou, E., Macdonald, E., Caslake, M., Stenvinkel, P., Shiels, P. G. (2016) Accelerated ageing and renal dysfunction links lower socioeconomic status and dietary phosphate intake. Aging, 8, pp. 1135-1149. (doi: 10.18632/aging.100948)

Woodling, N.S., Andreasson, K.I. (2016) Untangling the web: toxic and protective effects of neuroinflammation and PGE2 signaling in Alzheimer's disease. ACS Chemical Neuroscience, 7, pp. 454-463. (doi: 10.1021/acschemneuro.6b00016)

Bulteel, N., McGuinness, D., Shiels, P. G., McLauchlan, J. (2016) Circulating micrornas as biomarkers for disease progression in chronic hepatitis C virus infection.

Scialò, F., Sriram, A., Fernández-Ayala, D., Gubina, N., Lõhmus, M., Nelson, G., Logan, A., Cooper, H. M., Navas, P., Enríquez, J. A., Murphy, M. P., Sanz, A. (2016) Mitochondrial ROS produced via reverse electron transport extend animal lifespan. Cell Metabolism, 23, pp. 725-734. (doi: 10.1016/j.cmet.2016.03.009)

Modjtahedi, N., Tokatlidis, K., Dessen, P., Kroemer, G. (2016) Mitochondrial proteins containing coiled-coil-helix-coiled-coil-helix (CHCH) domains in health and disease. Trends in Biochemical Sciences, 41, pp. 245-260. (doi: 10.1016/j.tibs.2015.12.004)

Whalen, H., Shiels, P., Littlejohn, M., Clancy, M. (2016) A novel rodent model of severe renal ischemia reperfusion injury. Renal Failure, 38, pp. 1694-1701. (doi: 10.3109/0886022x.2016.1144024)

Regan, J. C., Khericha, M., Dobson, A. J., Bolukbasi, E., Rattanavirotkul, N., Partridge, L. (2016) Sex difference in pathology of the ageing gut mediates the greater response of female lifespan to dietary restriction. eLife, 5, (doi: 10.7554/eLife.10956)

Chaston, J. M., Dobson, A. J., Newell, P. D., Douglas, A. E. (2016) Host genetic control of the microbiota mediates the Drosophila nutritional phenotype. Applied and Environmental Microbiology, 82, pp. 671-679. (doi: 10.1128/AEM.03301-15)

McGuinness, D., Leierer, J., Shapter, O., Mohammed, S., Gingell-Littlejohn, M., Kingsmore, D. B., Little, A.-M., Kerschbaum, J., Schneeberger, S., Maglione, M., Nadalin, S., Wagner, S., Königsrainer, A., Aitken, E., Whalen, H., Clancy, M., McConnachie, A., Koppelstaetter, C., Stevenson, K. S., Shiels, P. G. (2016) Identification of molecular markers of delayed graft function based on the regulation of biological ageing. PLoS ONE, 11, (doi: 10.1371/journal.pone.0146378)

Stenvinkel, P., Kooman, J. P., Shiels, P. G. (2016) Nutrients and ageing: what can we learn about ageing interactions from animal biology? Current Opinion in Clinical Nutrition and Metabolic Care, 19, pp. 19-25. (doi: 10.1097/MCO.0000000000000234)

Khojah, S. M., McGuinness, D., Shiels, P. G., Payne, A. (2016) Differential Pattern of Global DNA Methylation at Two Time Points in Brain Regions of AS/AGU Rats Compared with AS Rats. (doi: 10.1142/9781783269150_0051)

Kato, S., Shiels, P. G., McGuinness, D., Lindholm, B., Stenvinkel, P., Nordfors, L., Qureshi, A. R., Yuzawa, Y., Matsuo, S., Maruyama, S. (2016) Telomere attrition and elongation after dialysis therapy initiation in patients with end-stage renal disease. Blood Purification, 41, pp. 25-33. (doi: 10.1159/000440971)

2015

Curley, H., McGuinness, D., Kinsella, J., Quasim, T., Shiels, P. (2015) Intensive care unit outcomes based on telomere length as a biomarker of ageing: a pilot study. British Journal of Anaesthesia, 115, pp. 950-962. (doi: 10.1093/bja/aev116)

Kooman, J. P., Shiels, P. G., Stenvinkel, P. (2015) Premature aging in chronic kidney disease and chronic obstructive pulmonary disease. Current Opinion in Clinical Nutrition and Metabolic Care, 18, pp. 528-534. (doi: 10.1097/mco.0000000000000218)

Gerhold, J. M., Cansiz-Arda, Ş., Lõhmus, M., Engberg, O., Reyes, A., van Rennes, H., Sanz, A., Holt, I. J., Cooper, H. M., Spelbrink, J. N. (2015) Human mitochondrial DNA-protein complexes attach to a cholesterol-rich membrane structure. Scientific Reports, 5, (doi: 10.1038/srep15292)

Martin-Ruiz, C. M., Baird, D., Roger, L., Boukamp, P., Krunic, D., Cawthon, R., Dokter, M. M., Van Der Harst, P., Bekaert, S., De Meyer, T., Roos, G., Svenson, U., Codd, V., Samani, N. J., McGlynn, L., Shiels, P. G., Pooley, K. A., Dunning, A. M., Cooper, R., Wong, A., Kingston, A., Von Zglinicki, T. (2015) Is Southern blotting necessary to measure telomere length reproducibly? Authors’ Response to: Commentary: The reliability of telomere length measurements. International Journal of Epidemiology, 44, pp. 1686-1687. (doi: 10.1093/ije/dyv169)

Rovenko, B. M., Kubrak, O. I., Gospodaryov, D. V., Yurkevych, I. S., Sanz, A., Lushchak, O. V., Lushchak, V. I. (2015) Restriction of glucose and fructose causes mild oxidative stress independently of mitochondrial activity and reactive oxygen species in Drosophila melanogaster. Comparative Biochemistry and Physiology. Part A: Molecular and Integrative Physiology, 187, pp. 27-39. (doi: 10.1016/j.cbpa.2015.04.012)

Syrjänen, L., Valanne, S., Kuuslahti, M., Tuomela, T., Sriram, A., Sanz, A., Jacobs, H. T., Rämet, M., Parkkila, S. (2015) β carbonic anhydrase is required for female fertility in Drosophila melanogaster. Frontiers in Zoology, 12, (doi: 10.1186/s12983-015-0111-3)

Rovenko, B. M., Kubrak, O. I., Gospodaryov, D. V., Perkhulyn, N. V., Yurkevych, I. S., Sanz, A., Lushchak, O. V., Lushchak, V. I. (2015) High sucrose consumption promotes obesity whereas its low consumption induces oxidative stress in Drosophila melanogaster. Journal of Insect Physiology, 79, pp. 42-54. (doi: 10.1016/j.jinsphys.2015.05.007)

Johansson, J.U., Woodling, N.S., Shi, J., Andreasson, K.I. (2015) Inflammatory cyclooxygenase activity and PGE2 signaling in models of alzheimer’s disease. Current Immunology Reviews, 11, pp. 125-131. (doi: 10.2174/1573395511666150707181414)

Dutta, D., Dobson, A. J., Houtz, P. L., Gläßer, C., Revah, J., Korzelius, J., Patel, P. H., Edgar, B. A., Buchon, N. (2015) Regional cell-specific transcriptome mapping reveals regulatory complexity in the adult Drosophila midgut. Cell Reports, 12, pp. 346-358. (doi: 10.1016/j.celrep.2015.06.009)

McGlynn, L. M., McCluney, S., Jamieson, N. B., Thomson, J., MacDonald, A. I., Oien, K., Dickson, E. J., Carter, C. R., McKay, C. J., Shiels, P. G. (2015) SIRT3 & SIRT7: potential novel biomarkers for determining outcome in pancreatic cancer patients. PLoS ONE, 10, (doi: 10.1371/journal.pone.0131344)

Johansson, J.U., Woodling, N.S., Brown, H.D., Wang, Q., Andreasson, K.I. (2015) Microarray analysis of the in vivo response of microglia to Aβ peptides in mice with conditional deletion of the prostaglandin EP2 receptor. Genomics Data, 5, pp. 268-271. (doi: 10.1016/j.gdata.2015.06.011)

Hangen, E. et al. (2015) Interaction between AIF and CHCHD4 regulates respiratory chain biogenesis. Molecular Cell, 58, pp. 1001-1014. (doi: 10.1016/j.molcel.2015.04.020)

Dobson, A. J., Chaston, J. M., Newell, P. D., Donahue, L., Hermann, S. L., Sannino, D. R., Westmiller, S., Wong, A. C.-N., Clark, A. G., Lazzaro, B. P., Douglas, A. E. (2015) Corrigendum: Host genetic determinants of microbiota-dependent nutrition revealed by genome-wide analysis of Drosophila melanogaster. Nature Communications, 6, (doi: 10.1038/ncomms8296)

Dobson, A. J., Chaston, J. M., Newell, P. D., Donahue, L., Hermann, S. L., Sannino, D. R., Westmiller, S., Wong, A. C.-N., Clark, A. G., Lazzaro, B. P., Douglas, A. E. (2015) Host genetic determinants of microbiota-dependent nutrition revealed by genome-wide analysis of Drosophila melanogaster. Nature Communications, 6, (doi: 10.1038/ncomms7312)

Rovenko, B. M., Perkhulyn, N. V., Gospodaryov, D. V., Sanz, A., Lushchak, O. V., Lushchak, V. I. (2015) High consumption of fructose rather than glucose promotes a diet-induced obese phenotype in Drosophila melanogaster. Comparative Biochemistry and Physiology. Part A: Molecular and Integrative Physiology, 180, pp. 75-85. (doi: 10.1016/j.cbpa.2014.11.008)

Shiels, P., Ritzau-Reid, K. (2015) Biological ageing, inflammation and nutrition: how might they impact on systemic sclerosis? Current Aging Science, 8, pp. 123-130. (doi: 10.2174/187460980801150727110353)

Ochoa, E. et al. (2015) Confirmation of CCR6 as a risk factor for anti-topoisomerase I antibodies in systemic sclerosis. Clinical and Experimental Rheumatology, 33, pp. S31-S35.

Martin-Ruiz, C. M., Baird, D., Roger, L., Boukamp, P., Krunic, D., Cawthon, R., Dokter, M. M., Van Der Harst, P., Bekaert, S., De Meyer, T., Roos, G., Svenson, U., Codd, V., Samani, N. J., McGlynn, L., Shiels, P. G., Pooley, K. A., Dunning, A. M., Cooper, R., Wong, A., Kingston, A., Von Zglinicki, T. (2015) Reproducibility of telomere length assessment: Authors’ Response to Damjan Krstajic and Ljubomir Buturovic. International Journal of Epidemiology, 44, pp. 1739-1741. (doi: 10.1093/ije/dyv170)

Scialò, F., Sriram, A., Naudí, A., Ayala, V., Jové, M., Pamplona, R., Sanz, A. (2015) Target of rapamycin activation predicts lifespan in fruit flies. Cell Cycle, 14, pp. 2949-2958. (doi: 10.1080/15384101.2015.1071745)

Mordas, A., Tokatlidis, K. (2015) The MIA pathway: a key regulator of mitochondrial oxidative protein folding and biogenesis. Accounts of Chemical Research, 48, pp. 2191-2199. (doi: 10.1021/acs.accounts.5b00150)

Postgraduate research students

Refine By

Background 

The global population demographic comprising those aged 65 years and over is increasing and expected to continue to grow over the remainder of the 21st century. By 2050, the population aged >65 years in the European Union is projected to reach ~130 million.

Despite the associated increase in human life span over the preceding 150 years, this has not been matched by a similar increase in health span (i.e. years of disease free living). Consequently, ageing populations present with more age-/lifestyle related diseases, including chronic kidney disease (CKD), cardiovascular disease (CVD), type 2 diabetes, osteoporosis, cancer, and neurodegenerative disorders.

This ‘diseasome of ageing’ has major clinical and socio-economic impacts on public health care across countries, raising challenges in terms of health equality and sustainability for healthcare provision. 

As a direct consequence, the UK Government has identified ‘Ageing Society’ as one of four Grand Challenges, with a mission to (i) improve health span by 5 years by 2035 and (ii) narrow the health span disparity gap between the deprived and affluent. Additionally, research into healthy (normative) ageing is at the forefront of UKRI’s Strategy 2022-2027.

The UKRI Strategic Theme, ‘Securing Better Health, Ageing, and Wellbeing’, is designed to improve people’s health span and promote wellbeing, in order to maintain prosperous, productive and resilient communities, both nationally and internationally 

The Science 

Ageing results in a loss of physical and physiological capability over time and increased susceptibility to disease. It results in loss of adaptive homeostasis, physiological dysfunction and eventually death. The rate of loss varies between individuals, between tissues and organs within the same individual and between different stages of the life course. Furthermore, ageing is subject to antagonistic pleiotropy and as such its effects on physical and physiological capability can be mitigated differentially. 

Ageing can be hallmarked by cellular and molecular features common across taxa (i.e. yeast, flies, worms, mice and humans), that reflect both accumulated molecular damage and the dysregulation of cellular processes during ageing. Whether these hallmarks of ageing function independently, cumulatively or synergistically with exposome factors (i.e. lifecourse biotic and abiotic exposures), remains to be understood. 

Our approach 

We have developed a cross disciplinary approach to understand determinants of healthy ageing, from basic mechanisms to senotherapies, designed to provide a holistic approach to healthy ageing. This comprises a translation circle, incorporating studies of (i) human ageing in the general population; (ii) basic mechanisms of the ageing in model organisms; (iii) the molecular basis of diseases of ageing; (iv) senotherapies; (vi) interventions in diseases of ageing and frailty. 

Research areas 

+++

The Exposome and Ageing 

Paul Shiels is Professor of Geroscience at the University of Glasgow. He is a a founder member of the Glasgow Geroscience Group (G3). He has established a reputation in the biology of ageing and is author of over 200 peer reviewed publications and a number of Patents in this sector.  His h-index is 54 and he has over 11,800 citations. He has published in high impact journals including Nature, Cell, Nature Genetics, Nature Neuroscience, Nature Reviews in Nephrology, Cell Reports J. Int Medicine, and PLoS Genetics, Aging Cell, Ann. Rheum. Dis, and Annals of Surgery. Prof. Shiels was a pioneer of telomere cloning and subsequently was the first to apply telomere analysis in cloned animals, including Dolly the sheep. He was subsequently the first to report on socioeconomic position (SEP) and a range of nutritional factors affecting epigenetic influences on ageing and health.  Additionally, he was the first to describe links between socio-economic position, the microbiome and ageing.

His current research portfolio comprises investigation and application of novel senotherapies and how the microbiome impacts on age related health. Paul has acted as an expert on the Biology of Ageing on a number of national policy advising consortia including providing evidence to UKXIRA and the All Party Parliamentary Group on Longevity (2019,2021). He is Chair of the FWO (Belgium) Med 6 Fellowship Funding Panel and is a panel member for the UK Research Partnership Investment Fund (UKRPIF). He sits on the Editorial Advisory Board for Aging Cell, Frontiers in Aging, Current Aging Biology and holds the honour of being Chair of the  Scientific Advisory Board of the British Society for Research on Ageing. He has sat on the International Advisory Board for the Nobel Symposium 2019 and was a Plenary Presenter at the Nobel Symposium 2019. Paul has acted as CSO for Pathfinder Cell Therapy PLC and sat on the Scientific Advisory Boards of TC Biopharm and 4D Pharma and acts as a consultant for a range of Pharma companies. He has a proven track record in public dissemination of his research, including the provision of expert commentary for the BBC and ABC TV networks and as a Panellist at the Edinburgh International Science Festival and the Edinburgh International Book Festival.  

---

+++

The Mitochondrion and Ageing 

Professor Alberto Sanz has dedicated his professional life to unravelling the complex biology of ageing, emphasising developing therapies aimed at delaying, preventing, or even reversing the process and its related diseases. After earning his PhD in Biology from the Complutense University of Madrid and receiving additional training in Poland and the United States, he pursued a postdoctoral position in Finland, supported by a distinguished long-term EMBO fellowship. His extraordinary accomplishments were recognised with a Starting Grant from the European Research Council, facilitating the establishment of his own laboratory. During his tenure in Finland, the Finnish Academy honoured him with a Research Academy Fellowship. His journey continued at Newcastle University, where he excelled and was promoted to Chair of Biology of Ageing five years after joining the institution. His ground-breaking research also secured him a prestigious Senior Research Fellowship from the Wellcome Trust while he was at Newcastle.

Since 2020, Professor Sanz holds is a Lord Kelvin/Adam Smith Professor at the University of Glasgow. His laboratory, currently supported by the BBSRC, Wellcome Trust, and the University of Glasgow, undertakes essential research into the complex relationship between mitochondria and cellular ageing. Specifically, his work explores how mitochondrial signals contribute to cellular homeostasis and why this critical function deteriorates with ageing. Professor Sanz's innovative research has appeared in prestigious scientific journals such as Cell Metabolism, Nature Communications, Developmental Cell, EMBO Molecular Medicine and the Proceedings of the National Academy of Sciences, thanks to strategic partnerships with research groups worldwide. The impact of Professor Sanz's work is profound; his pioneering contributions to the field of ageing biology hold the potential to improve the quality of life by advocating for not merely longer but healthier lives.

---

+++

Microbiota, diet, signalling and ageing

Nutrition affects all aspects of animal function, determined by the interplay of diet and gut microbiota. We have known for almost a century that nutrition can be altered to promote healthy ageing, which is an attractive approach to promote general health in the population. There is now also abundant evidence that reduced nutrient signalling also promotes healthy ageing. Yet we still lack systems-level understanding of how nutrition alters organismal function. This information is key to (A) alleviating the significant biological costs of interventions (e.g. compromised reproduction, immunity, growth), and (B) correctly targeting interventions to match individual need.

We take a multi-level approach, scaling from molecules to populations, to understand mechanistically how nutrition and signalling affect ageing, variation in response to these interventions, and mechanisms of variation. We use Drosophila as a discovery vehicle to identify fundamental processes that are conserved throughout animal life; generating hypotheses that we then test in other systems. We are particularly interested in gut microbiota, identifying host nutritional processes that we expect are controlled by bacteria throughout animals. Drosophila we can control all aspects of the fly’s microbial and dietary environment, and then conduct powerful tissue-specific molecular genetics, allowing us to identify precisely how cell-type-specific molecular function is regulated by microbial or nutritional alterations, and the consequences for ageing at a population level.  

---

+++

Ageing and neurodegenerative diseases  

Dr Nathan Woodling is a Lecturer whose work aims to untangle the molecular and cellular processes of brain ageing that contribute to functional decline and neurodegenerative disease susceptibility. Dr Woodling completed his PhD in 2013 at Stanford University, where his work helped define how inflammatory signalling pathways in microglia contributed to the types of neurotoxicity seen in Alzheimer’s disease. He then moved to the UCL Institute of Healthy Ageing for his postdoctoral work, which investigated how both glial and neuronal cells can modulate ageing of the nervous system through distinct signalling pathways.

In 2019, he was awarded an Alzheimer’s Society Junior Fellowship, which supported his investigation of age-related changes in glia that predispose the nervous system to the types of damage that occur in Alzheimer’s disease and other dementia-causing diseases. In 2022, Dr Woodling moved to Glasgow to start his own research group in the School of Molecular Biosciences. His lab now investigates cellular signalling pathways that could be targeted by new or repurposed treatment strategies to maintain healthy function and reduce disease susceptibility during ageing.

---